Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 26
Filtrar
1.
J Biol Chem ; 294(12): 4546-4571, 2019 03 22.
Artículo en Inglés | MEDLINE | ID: mdl-30696771

RESUMEN

Na+-H+ exchanger regulatory factor-1 (NHERF1) is a PDZ protein that scaffolds membrane proteins, including sodium-phosphate co-transport protein 2A (NPT2A) at the plasma membrane. NHERF1 is a phosphoprotein with 40 Ser and Thr residues. Here, using tandem MS analysis, we characterized the sites of parathyroid hormone (PTH)-induced NHERF1 phosphorylation and identified 10 high-confidence phosphorylation sites. Ala replacement at Ser46, Ser162, Ser181, Ser269, Ser280, Ser291, Thr293, Ser299, and Ser302 did not affect phosphate uptake, but S290A substitution abolished PTH-dependent phosphate transport. Unexpectedly, Ser290 was rapidly dephosphorylated and rephosphorylated after PTH stimulation, and we found that protein phosphatase 1α (PP1α), which binds NHERF1 through a conserved VxF/W PP1 motif, dephosphorylates Ser290 Mutating 257VPF259 eliminated PP1 binding and blunted dephosphorylation. Tautomycetin blocked PP1 activity and abrogated PTH-sensitive phosphate transport. Using fluorescence lifetime imaging (FLIM), we observed that PTH paradoxically and transiently elevates intracellular phosphate. Added phosphate blocked PP1α-mediated Ser290 dephosphorylation of recombinant NHERF1. Hydrogen-deuterium exchange MS revealed that ß-sheets in NHERF1's PDZ2 domain display lower deuterium uptake than those in the structurally similar PDZ1, implying that PDZ1 is more cloistered. Dephosphorylated NHERF1 exhibited faster exchange at C-terminal residues suggesting that NHERF1 dephosphorylation precedes Ser290 rephosphorylation. Our results show that PP1α and NHERF1 form a holoenzyme and that a multiprotein kinase cascade involving G protein-coupled receptor kinase 6A controls the Ser290 phosphorylation status of NHERF1 and regulates PTH-sensitive, NPT2A-mediated phosphate uptake. These findings reveal how reversible phosphorylation modifies protein conformation and function and the biochemical mechanisms underlying PTH control of phosphate transport.


Asunto(s)
Hormona Paratiroidea/fisiología , Fosfatos/metabolismo , Fosfoproteínas/metabolismo , Intercambiadores de Sodio-Hidrógeno/metabolismo , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIa/fisiología , Secuencia de Aminoácidos , Cristalografía por Rayos X , Furanos/farmacología , Células HEK293 , Humanos , Transporte Iónico/fisiología , Lípidos/farmacología , Fosfoproteínas/química , Fosforilación , Conformación Proteica , Receptores de Neuropéptido Y/antagonistas & inhibidores , Receptores de Neuropéptido Y/metabolismo , Serina/metabolismo , Intercambiadores de Sodio-Hidrógeno/química
2.
Physiol Rep ; 6(12): e13715, 2018 06.
Artículo en Inglés | MEDLINE | ID: mdl-29924459

RESUMEN

Mutations in SLC34A1, encoding the proximal tubular sodium-phosphate transporter NaPi-IIa, may cause a range of clinical phenotypes including infantile hypercalcemia, a proximal renal Fanconi syndrome, which are typically autosomal recessive, and hypophosphatemic nephrolithiasis, which may be an autosomal dominant trait. Here, we report two patients with mixed clinical phenotypes, both with metabolic acidosis, hyperphosphaturia, and renal stones. Patient A had a single heterozygous pathogenic missense mutation (p.I456N) in SLC34A1, consistent with the autosomal dominant pattern of renal stone disease in this family. Patient B, with an autosomal recessive pattern of disease, was compound heterozygous for SLC34A1 variants; a missense variant (p.R512C) together with a relatively common in-frame deletion p.V91A97del7 (91del7). Xenopus oocyte and renal (HKC-8) cell line transfection studies of the variants revealed limited cell surface localization, consistent with trafficking defects. Co-expression of wild-type and I456N and 91del7 appeared to cause intracellular retention in HKC-8, whereas the R512C mutant had a less dominant effect. Expression in Xenopus oocytes failed to demonstrate a significant dominant negative effect for I456N and R512C; however, a negative impact of 91del7 on [32 P]phosphate transport was found. In conclusion, we have investigated pathogenic alleles of SLC34A1 which contribute to both autosomal dominant and autosomal recessive renal stone disease.


Asunto(s)
Mutación , Nefrolitiasis/genética , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIa/genética , Adulto , Simulación por Computador , Humanos , Hipofosfatemia/genética , Hipofosfatemia/metabolismo , Lactante , Masculino , Nefrolitiasis/metabolismo , Fenotipo , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIa/metabolismo , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIa/fisiología
3.
Toxicol Pathol ; 45(7): 904-910, 2017 10.
Artículo en Inglés | MEDLINE | ID: mdl-29096595

RESUMEN

Fibroblast growth factor-23 (FGF23) is a bone-derived hormone, mainly produced by osteoblasts and osteocytes in response to increased extracellular phosphate and circulating vitamin D hormone. Endocrine FGF23 signaling requires co-expression of the ubiquitously expressed FGF receptor 1 (FGFR1) and the co-receptor α-Klotho (Klotho). In proximal renal tubules, FGF23 suppresses the membrane expression of the sodium-phosphate cotransporters Npt2a and Npt2c which mediate urinary reabsorption of filtered phosphate. In addition, FGF23 suppresses proximal tubular expression of 1α-hydroxylase, the key enzyme responsible for vitamin D hormone production. In distal renal tubules, FGF23 signaling activates with-no-lysine kinase 4, leading to increased renal tubular reabsorption of calcium and sodium. Therefore, FGF23 is not only a phosphaturic but also a calcium- and sodium-conserving hormone, a finding that may have important implications for the pathophysiology of chronic kidney disease. Besides these endocrine, Klotho-dependent functions of FGF23, FGF23 is also an auto-/paracrine suppressor of tissue-nonspecific alkaline phosphatase transcription via Klotho-independent FGFR3 signaling, leading to local inhibition of mineralization through accumulation of pyrophosphate. In addition, FGF23 may target the heart via an FGFR4-mediated Klotho-independent signaling cascade. Taken together, there is emerging evidence that FGF23 is a pleiotropic hormone, linking bone with several other organ systems.


Asunto(s)
Huesos/fisiología , Factores de Crecimiento de Fibroblastos/fisiología , Glucuronidasa/fisiología , Comunicación Autocrina , Calcificación Fisiológica , Sistema Cardiovascular , Factor-23 de Crecimiento de Fibroblastos , Humanos , Inmunomodulación , Túbulos Renales Proximales/fisiología , Proteínas Klotho , Comunicación Paracrina , Fosfatos/fisiología , Receptor Tipo 1 de Factor de Crecimiento de Fibroblastos/fisiología , Receptor Tipo 3 de Factor de Crecimiento de Fibroblastos/fisiología , Receptor Tipo 4 de Factor de Crecimiento de Fibroblastos/fisiología , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIa/fisiología , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIc/fisiología
4.
Clin Calcium ; 26(2): 284-94, 2016 Feb.
Artículo en Japonés | MEDLINE | ID: mdl-26813509

RESUMEN

Serum level of phosphate is regulated by the kidney, especially proximal tubule. The transcellular transport of phosphate in the proximal tubule is mediated via Na dependent transporters, i.e., NPT2a and NPT2b at the luminal membrane, and unknown channel at the basolateral side. The transport of phosphate via NPT2a and NPT2b is further regulated by factors, such as PTH, FGF23, and 1,25(OH)(2)D. Several hereditary diseases that cause hypophoshatemia specically are known. In addition, dysfunction of proximal tubule may develop Fanconi syndrome, which also causes hypherphosphaturia. In this section, I describe the renal mechanisms of phosphate handling and the causes of hypophosphatemia along with its treatment.


Asunto(s)
Hipofosfatemia/etiología , Hipofosfatemia/metabolismo , Túbulos Renales Proximales/metabolismo , Fosfatos/metabolismo , Administración Oral , Calcitriol/fisiología , Canales de Cloruro , Enfermedad de Dent/etiología , Enfermedad de Dent/genética , Enfermedad de Dent/metabolismo , Síndrome de Fanconi/etiología , Síndrome de Fanconi/metabolismo , Factor-23 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/fisiología , Humanos , Hipofosfatemia/terapia , Enfermedades Mitocondriales , Síndrome Oculocerebrorrenal , Hormona Paratiroidea/fisiología , Monoéster Fosfórico Hidrolasas , Compuestos de Fósforo/administración & dosificación , Compuestos de Fósforo/uso terapéutico , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIa/fisiología , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIc/fisiología , Vitamina D/administración & dosificación
5.
Biosci Biotechnol Biochem ; 80(3): 510-3, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26460967

RESUMEN

Vitamin B12 deficiency is a risk factor for bone disorders via mechanisms not fully understood. In this study, an increase in serum inorganic phosphorus (Pi) concentrations was associated with a vitamin B12 deficiency. Napi2a, a renal cotransporter for Pi reabsorption, accumulated on plasma membranes in a vitamin B12 deficiency suggests that vitamin B12 plays an important role in Pi homeostasis.


Asunto(s)
Resorción Ósea , Riñón/metabolismo , Osteoclastos/patología , Fosfatos/metabolismo , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIa/fisiología , Deficiencia de Vitamina B 12/fisiopatología , Animales , Homeostasis , Ratas
6.
J Am Soc Nephrol ; 25(4): 761-72, 2014 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-24262791

RESUMEN

Marked hypophosphatemia is common after major hepatic resection, but the pathophysiologic mechanism remains unknown. We used a partial hepatectomy (PH) rat model to investigate the molecular basis of hypophosphatemia. PH rats exhibited hypophosphatemia and hyperphosphaturia. In renal and intestinal brush-border membrane vesicles isolated from PH rats, Na(+)-dependent phosphate (Pi) uptake decreased by 50%-60%. PH rats also exhibited significantly decreased levels of renal and intestinal Na(+)-dependent Pi transporter proteins (NaPi-IIa [NaPi-4], NaPi-IIb, and NaPi-IIc). Parathyroid hormone was elevated at 6 hours after PH. Hyperphosphaturia persisted, however, even after thyroparathyroidectomy in PH rats. Moreover, DNA microarray data revealed elevated levels of nicotinamide phosphoribosyltransferase (Nampt) mRNA in the kidney after PH, and Nampt protein levels and total NAD concentration increased significantly in the proximal tubules. PH rats also exhibited markedly increased levels of the Nampt substrate, urinary nicotinamide (NAM), and NAM catabolites. In vitro analyses using opossum kidney cells revealed that NAM alone did not affect endogenous NaPi-4 levels. However, in cells overexpressing Nampt, the addition of NAM led to a marked decrease in cell surface expression of NaPi-4 that was blocked by treatment with FK866, a specific Nampt inhibitor. Furthermore, FK866-treated mice showed elevated renal Pi reabsorption and hypophosphaturia. These findings indicate that hepatectomy-induced hypophosphatemia is due to abnormal NAM metabolism, including Nampt activation in renal proximal tubular cells.


Asunto(s)
Hepatectomía/efectos adversos , Hipofosfatemia/etiología , Riñón/metabolismo , Acrilamidas/farmacología , Animales , Masculino , Ratones , Ratones Endogámicos C57BL , NAD/metabolismo , Niacinamida/metabolismo , Nicotinamida Fosforribosiltransferasa/fisiología , Paratiroidectomía , Piperidinas/farmacología , Ratas , Ratas Wistar , Proteínas Cotransportadoras de Sodio-Fosfato/fisiología , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIa/fisiología
7.
J Med Invest ; 60(1-2): 27-34, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23614908

RESUMEN

Type IIa sodium-dependent phosphate transporter (NaPi-IIa) is responsible for renal phosphate reabsorption and maintenance of systemic phosphate homeostasis in mammals. Macromolecular complex formation of NaPi-IIa with sodium-proton exchanger related factor-1 (NHERF-1) and ezrin is important for apical membrane localization in the proximal tubular cells. Here, we investigated the interactions of the ezrin phosphomimetic mutation of serine to aspartic acid at 249 with NHERF-1 and the inhibition of apical membrane localization of NaPi-IIa. In vitro phosphorylation analysis revealed that serine 249 of human ezrin serves as a phosphorylation site for protein kinase A. The N-terminal half of ezrin had a dominant negative effect on the phosphate transport activity and inhibited the apical localization of NaPi-IIa in renal proximal tubular cells. We found that the phosphomimetic S249D mutant interfered with the inhibitory effects of the dominant negative mutant on the transport and localization of NaPi-IIa. The S249D mutant also inhibited the interaction with NHERF-1. Therefore, serine 249 of ezrin can play important roles in the regulation of the complex formation and membrane localization of NaPi-IIa.


Asunto(s)
Proteínas del Citoesqueleto/fisiología , Túbulos Renales Proximales/metabolismo , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIa/fisiología , Actinas/metabolismo , Animales , Células COS , Células Cultivadas , Chlorocebus aethiops , Proteínas Quinasas Dependientes de AMP Cíclico/fisiología , Proteínas del Citoesqueleto/química , Fosfoproteínas/metabolismo , Fosforilación , Serina , Intercambiadores de Sodio-Hidrógeno/metabolismo
8.
Annu Rev Physiol ; 75: 535-50, 2013.
Artículo en Inglés | MEDLINE | ID: mdl-23398154

RESUMEN

Plasma phosphate concentration is maintained within a relatively narrow range by control of renal reabsorption of filtered inorganic phosphate (P(i)). P(i) reabsorption is a transcellular process that occurs along the proximal tubule. P(i) flux at the apical (luminal) brush border membrane represents the rate-limiting step and is mediated by three Na(+)-dependent P(i) cotransporters (members of the SLC34 and SLC20 families). The putative proteins responsible for basolateral P(i) flux have not been identified. The transport mechanism of the two kidney-specific SLC34 proteins (NaPi-IIa and NaPi-IIc) and of the ubiquitously expressed SLC20 protein (PiT-2) has been studied by heterologous expression to reveal important differences in kinetics, stoichiometry, and substrate specificity. Studies on the regulation of the abundance of the respective proteins highlight significant differences in the temporal responses to various hormonal and nonhormonal factors that can influence P(i) homeostasis. The phenotypes of mice deficient in NaPi-IIa and NaPi-IIc indicate that NaPi-IIa is responsible for most P(i) renal reabsorption. In contrast, in the human kidney, NaPi-IIc appears to have a relatively greater role. The physiological relevance of PiT-2 to P(i) reabsorption remains to be elucidated.


Asunto(s)
Homeostasis/fisiología , Túbulos Renales Proximales/fisiología , Proteínas de Transporte de Fosfato/fisiología , Animales , Humanos , Ratones , Fosfatos/metabolismo , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIa/fisiología , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIc/fisiología
9.
Cell Physiol Biochem ; 28(2): 251-8, 2011.
Artículo en Inglés | MEDLINE | ID: mdl-21865732

RESUMEN

Klotho, a transmembrane protein, protease and hormone has been shown to exert a profound effect on phosphate metabolism. Klotho overexpression lowers and Klotho deficiency increases the plasma phosphate concentration, effects in part attributed to an inhibitory effect of Klotho on the formation of 1,25-dihydroxycholecalciferol (1,25(OH) (2)D(3)), the active form of Vitamin D. Beyond that Klotho has been shown to decrease renal tubular phosphate transport more directly. The influence of Klotho on the plasma phosphate concentration contributes to the profound effect of Klotho on ageing and life span. The present study explored whether Klotho influences the major renal tubular (NaPi-IIa) and the major intestinal (NaPi-IIb) phosphate transporters. For functional analysis NaPi-IIa or NaPi-IIb were expressed in Xenopus oocytes both, without or with additional coexpression of Klotho and electrogenic phosphate transport was estimated from the phosphate-induced current (Ip). According to RT-PCR Klotho is expressed in the murine kidney and intestine. Coexpression of Klotho decreased Ip in both NaPi-IIa- and NaPi-IIb-expressing oocytes. Klotho decreased the maximal Ip without appreciably affecting the concentration required for halfmaximal Ip. Treatment of NaPi-IIa- or NaPi-IIb-expressing oocytes with Klotho protein similarly decreased Ip. In conclusion, Klotho down regulates both, renal (NaPi-IIa) and intestinal (NaPi-IIb) phosphate transporters.


Asunto(s)
Regulación hacia Abajo , Glucuronidasa/metabolismo , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIa/metabolismo , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIb/metabolismo , Animales , Calcifediol/farmacología , Regulación hacia Abajo/efectos de los fármacos , Glucuronidasa/genética , Mucosa Intestinal/metabolismo , Intestinos/efectos de los fármacos , Riñón/efectos de los fármacos , Riñón/metabolismo , Proteínas Klotho , Ratones , Oocitos/efectos de los fármacos , Oocitos/metabolismo , Oocitos/fisiología , Fosfatos/metabolismo , Fosfatos/farmacología , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIa/genética , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIa/fisiología , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIb/genética , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIb/fisiología , Xenopus laevis
12.
Endocrinology ; 151(10): 4607-12, 2010 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-20685875

RESUMEN

Rickets is a growth plate abnormality observed in growing animals and humans. Rachitic expansion of the hypertrophic chondrocyte layer of the growth plate, in the setting of hypophosphatemia, is due to impaired apoptosis of these cells. Rickets is observed in humans and mice with X-linked hypophosphatemia that is associated with renal phosphate wasting secondary to elevated levels of fibroblast growth factor-23. Rickets is also seen in settings of impaired vitamin D action, due to elevated PTH levels that increase renal phosphate excretion. However, mice with hypophosphatemia secondary to ablation of the renal sodium-dependent phosphate transport protein 2a (Npt2a), have not been reported to develop rickets. Because activation of the mitochondrial apoptotic pathway by phosphate is required for hypertrophic chondrocyte apoptosis in vivo, investigations were undertaken to address this paradox. Analyses of the Npt2a null growth plate demonstrate expansion of the hypertrophic chondrocyte layer at 2 wk of age, with resolution of this abnormality by 5 wk of age. This is temporally associated with an increase in circulating levels of 1,25-dihydroxyvitamin D. To address whether the receptor-dependent actions of this steroid hormone are required for normalization of the growth plate phenotype, the Npt2a null mice were mated with mice lacking the vitamin D receptor or were rendered vitamin D deficient. These studies demonstrate that the receptor-dependent actions of 1,25-dihydroxyvitamin D are required for maintenance of a normal growth plate phenotype in the Npt2a null mice.


Asunto(s)
Placa de Crecimiento/crecimiento & desarrollo , Receptores de Calcitriol/fisiología , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIa/genética , Vitamina D/análogos & derivados , Animales , Calcio/sangre , Calcio/metabolismo , Femenino , Factor-23 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/sangre , Factores de Crecimiento de Fibroblastos/metabolismo , Placa de Crecimiento/efectos de los fármacos , Placa de Crecimiento/metabolismo , Hipofosfatemia/genética , Hipofosfatemia/metabolismo , Hipofosfatemia/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Hormona Paratiroidea/sangre , Hormona Paratiroidea/metabolismo , Fosfatos/sangre , Fosfatos/metabolismo , Receptores de Calcitriol/genética , Receptores de Calcitriol/metabolismo , Raquitismo/genética , Raquitismo/metabolismo , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIa/fisiología , Vitamina D/sangre , Vitamina D/metabolismo , Vitamina D/farmacología , Vitamina D/fisiología
13.
Urol Res ; 38(6): 429-38, 2010 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-20658131

RESUMEN

Nephrocalcinosis is the deposition of calcium salts in renal parenchyma and can be intratubular or interstitial. Animal model studies indicate that intratubular nephrocalcinosis is a result of increased urinary supersaturation. Urinary supersaturation with respect to calcium oxalate (CaOx) and calcium phosphate (CaP) are generally achieved at different locations in the renal tubules. As a result experimental induction of hyperoxaluria in animals with CaP deposits does not lead to growth of CaOx over CaP. Interstitial nephrocalcinosis has been seen in mice with lack of crystallization modulators Tamm-Horsfall protein and osteopontin. Sodium phosphate co-transporter or sodiumhydrogen exchanger regulator factor-1 null mice also produced interstitial nephrocalcinosis. Crystals plug the tubules by aggregating and attaching to the luminal cell surface. Structural features of the renal tubules also play a role in crystal retention. The crystals plugging the terminal collecting ducts when exposed to the metastable pelvic urine may promote the formation of stone.


Asunto(s)
Modelos Animales de Enfermedad , Nefrocalcinosis/etiología , Animales , Oxalato de Calcio/metabolismo , Fosfatos de Calcio/metabolismo , Riñón/metabolismo , Ratones , Osteopontina/fisiología , Ratas , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIa/fisiología , Porcinos , Uromodulina/fisiología
14.
Urol Res ; 38(4): 271-6, 2010 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-20665015

RESUMEN

Control of phosphate (P(i)) homeostasis is essential for many biologic functions and inappropriate low levels of P(i) in plasma have been suggested to associate with several pathological states, including renal stone formation and stone recurrence. P(i) homeostasis is achieved mainly by adjusting the renal reabsorption of P(i) to the body's requirements. This task is performed to a major extent by the Na/Pi cotransporter NaPi-IIa that is specifically expressed in the brush border membrane of renal proximal tubules. While the presence of tight junctions in epithelial cells prevents the diffusion and mixing of the apical and basolateral components, the location of a protein within a particular membrane subdomain (i.e., the presence of NaPi-IIa at the tip of the apical microvilli) often requires its association with scaffolding elements which directly or indirectly connect the protein with the underlying cellular cytoskeleton. NaPi-IIa interacts with the four members of the Na(+)/H(+) exchanger regulatory factor family as well as with the GABA(A)-receptor associated protein . Here we will discuss the most relevant findings regarding the role of these proteins on the expression and regulation of the cotransporter, as well as the impact that their absence has in P(i) homeostasis.


Asunto(s)
Riñón/metabolismo , Fosfatos/metabolismo , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIa/fisiología , Absorción , Animales , Homeostasis , Ratones , Ratones Noqueados , Proteínas Asociadas a Microtúbulos/metabolismo , Modelos Biológicos , Dominios PDZ/fisiología , Fosfoproteínas/metabolismo , Intercambiadores de Sodio-Hidrógeno/metabolismo
15.
J Med Invest ; 57(1-2): 95-108, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-20299748

RESUMEN

Phosphaturia has been documented following cadmium (Cd) exposure in both humans and experimental animals. The fibroblast growth factor 23 (FGF23)/klotho axis serves as an essential phosphate homeostasis pathway in the bone-kidney axis. In the present study, we investigated the effects of Cd on phosphate (Pi) homeostasis in mice. Following Cd injection into WT mice, plasma FGF23 concentration was significantly increased. Urinary Pi excretion levels were significantly higher in Cd-injected WT mice than in control group. Plasma Pi concentration decreased only slightly compared with control group. No change was observed in plasma parathyroid hormone and 1,25-dihydroxy vitamin D(3) in both group of mice. We observed a decrease in phosphate transport activity and also decrease in expression of renal phosphate transporter SLC34A3 [NaPi-IIc/NPT2c], but not SLC34A1 [NaPi-IIa/NPT2a]. Furthermore, we examined the effect of Cd on Npt2c in Npt2a-knockout (KO) mice which expresses Npt2c as a major NaPi co-transporter. Injecting Cd to Npt2aKO mice induced significant increase in plasma FGF23 concentration and urinary Pi excretion levels. Furthermore, we observed a decrease in phosphate transport activity and renal Npt2c expression in Cd-injected Npt2a KO mice. The present study suggests that hypophosphatemia induced by Cd may be closely associated with the FGF23/klotho axis.


Asunto(s)
Cadmio/toxicidad , Factores de Crecimiento de Fibroblastos/fisiología , Hipofosfatemia Familiar/inducido químicamente , Animales , Proteínas de la Matriz Extracelular/genética , Femenino , Factor-23 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Endopeptidasa Neutra Reguladora de Fosfato PHEX/genética , ARN Mensajero/análisis , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIa/análisis , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIa/fisiología
16.
Biochem J ; 427(1): 161-9, 2010 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-20088828

RESUMEN

The type IIa renal sodium-dependent phosphate (Na/Pi) co-transporter Npt2a is implicated in the control of serum phosphate levels. It has been demonstrated previously that renal Npt2a protein and its mRNA expression are both up-regulated by the thyroid hormone T3 (3,3',5-tri-iodothyronine) in rats. However, it has never been established whether the induction was mediated by a direct effect of thyroid hormones on the Npt2a promoter. To address the role of Npt2a in T3-dependent regulation of phosphate homoeostasis and to identify the molecular mechanisms by which thyroid hormones modulate Npt2a gene expression, mice were rendered pharmacologically hypo- and hyper-thyroid. Hypothyroid mice showed low levels of serum phosphate and a marked decrease in renal Npt2a protein abundance. Importantly, we also showed that Npt2a-deficient mice had impaired serum phosphate responsiveness to T3 compared with wild-type mice. Promoter analysis with a luciferase assay revealed that the transcriptional activity of a reporter gene containing the Npt2a promoter and intron 1 was dependent upon TRs (thyroid hormone receptors) and specifically increased by T3 in renal cells. Deletion analysis and EMSAs (electrophoretic mobility-shift assays) determined that there were unique TREs (thyroid-hormone-responsive elements) within intron 1 of the Npt2a gene. These results suggest that Npt2a plays a critical role as a T3-target gene, to control phosphate homoeostasis, and that T3 transcriptionally activates the Npt2a gene via TRs in a renal cell-specific manner.


Asunto(s)
Regulación de la Expresión Génica , Riñón/metabolismo , Fosfatos/metabolismo , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIa/fisiología , Transcripción Genética/efectos de los fármacos , Triyodotironina/farmacología , Animales , Células COS , Chlorocebus aethiops , Perros , Ensayo de Cambio de Movilidad Electroforética , Femenino , Células HeLa , Homeostasis , Humanos , Riñón/citología , Luciferasas/metabolismo , Masculino , Ratones , Ratones Noqueados , Regiones Promotoras Genéticas , Ratas , Receptores de Hormona Tiroidea/metabolismo , Elementos de Respuesta , Activación Transcripcional
17.
Pflugers Arch ; 458(1): 39-52, 2009 May.
Artículo en Inglés | MEDLINE | ID: mdl-18758808

RESUMEN

Homeostasis of inorganic phosphate (P(i)) is primarily an affair of the kidneys. Reabsorption of the bulk of filtered P(i) occurs along the renal proximal tubule and is initiated by apically localized Na(+)-dependent P(i) cotransporters. Tubular P(i) reabsorption and therefore renal excretion of P(i) is controlled by a number of hormones, including phosphatonins, and metabolic factors. In most cases, regulation of P(i) reabsorption is achieved by changing the apical abundance of Na(+)/Pi cotransporters. The regulatory mechanisms involve various signaling pathways and a number of proteins that interact with Na(+)/P(i) cotransporters.


Asunto(s)
Túbulos Renales Proximales/metabolismo , Fosfatos/metabolismo , Proteínas Cotransportadoras de Sodio-Fosfato/fisiología , Acidosis/fisiopatología , Animales , Colecalciferol/fisiología , Ritmo Circadiano , Dieta , Factor-23 de Crecimiento de Fibroblastos , Factores de Crecimiento de Fibroblastos/fisiología , Regulación de la Expresión Génica , Glucuronidasa/fisiología , Hormonas Esteroides Gonadales/fisiología , Homeostasis , Humanos , Transporte Iónico , Proteínas Klotho , Hormona Paratiroidea/fisiología , Fosfatos/orina , Fosfoproteínas/fisiología , Deficiencia de Potasio/fisiopatología , Transducción de Señal , Intercambiadores de Sodio-Hidrógeno/fisiología , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIa/fisiología , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIc/fisiología
18.
Am J Physiol Renal Physiol ; 294(5): F1109-15, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18337544

RESUMEN

The most common theories about the pathogenesis of idiopathic kidney stones consider precipitation of calcium phosphate (CaP) within the kidneys critical for the development of the disease. We decided to test the hypothesis that a CaP substrate can promote the deposition of calcium oxalate (CaOx) in the kidneys. Experimental hyperoxaluria was induced by feeding glyoxylate to male mice with knockout (KO) of NaP(i) IIa (Npt2a), a sodium-phosphate cotransporter. Npt2a KO mice are hypercalciuric and produce CaP deposits in their renal tubules. Experimental hyperoxaluria led to CaOx crystalluria in both the hypercalciuric KO mice and the normocalciuric control B6 mice. Only the KO mice produced CaOx crystal deposits in their kidneys, but the CaOx crystals deposited separately from the CaP deposits. Perhaps CaP deposits were not available for a CaOx overgrowth. These results also validate earlier animal model observations that showed that CaP substrate is not required for renal deposition of CaOx and that other factors, such as local supersaturation, may be involved. The absence of CaOx deposition in the B6 mice despite extreme hyperoxaluria also signifies the importance of both calcium and oxalate in the development of CaOx nephrolithiasis.


Asunto(s)
Oxalato de Calcio/metabolismo , Hipercalciuria/patología , Riñón/patología , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIa/genética , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIa/fisiología , Envejecimiento/metabolismo , Envejecimiento/fisiología , Animales , Conducta Animal/fisiología , Peso Corporal/fisiología , Cristalización , Concentración de Iones de Hidrógeno , Masculino , Ratones , Ratones Noqueados , Microscopía Electrónica de Rastreo , Mutagénesis , Adhesión en Parafina
19.
Kidney Int ; 73(10): 1141-50, 2008 May.
Artículo en Inglés | MEDLINE | ID: mdl-18305465

RESUMEN

Estrogen treatment causes significant hypophosphatemia in patients. To determine the mechanisms responsible for this effect, we injected ovariectomized rats with either 17beta-estradiol or vehicle for three days. Significant renal phosphate wasting and hypophosphatemia occurred in estrogen-treated rats despite a decrease in their food intake. The mRNA and protein levels of the renal proximal tubule sodium phosphate cotransporter (NaPi-IIa) were significantly decreased in estradiol-treated ad-libitum or pair-fed groups. Estrogen did not affect NaPi-III or NaPi-IIc expression. In ovariectomized and parathyroidectomized rats, 17beta-estradiol caused a significant decrease in NaPi-IIa mRNA and protein expression compared to vehicle. Estrogen receptor alpha isoform blocker significantly blunted the anorexic effect of 17beta-estradiol but did not affect the downregulation of NaPi-IIa. Our studies show that renal phosphate wasting and hypophosphatemia induced by estrogen are secondary to downregulation of NaPi-IIa in the proximal tubule. These effects are independent of food intake or parathyroid hormone levels and likely not mediated through the activation of estrogen receptor alpha subtype.


Asunto(s)
Regulación hacia Abajo , Estrógenos/fisiología , Hipofosfatemia/etiología , Fosfatos/metabolismo , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIa/fisiología , Animales , Estradiol/administración & dosificación , Túbulos Renales Proximales/metabolismo , Ratas
20.
Toxicol Sci ; 101(2): 254-62, 2008 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-17998273

RESUMEN

It has been suggested that uranium uptake and toxicity could be mediated by endocytosis and/or the type IIa sodium-dependent phosphate cotransporter (NaPi-IIa). The aim of this study was therefore to characterize in vitro the role of these two cellular mechanisms in the uptake and toxicity of low (200-3200 nM) and high (0.5 and 0.8 mM) concentrations of uranium, respectively. At low concentrations, uranium uptake in LLC-PK(1) cells was saturable (V(max) = 3.09 +/- 0.22 ng/mg protein) and characterized by a K(0.5) of 1022 +/- 63 nM and a Hill coefficient of 3.0 +/- 0.4. The potential involvement of endocytosis and NaPi-IIa in the uptake of uranium was assessed by the use of various drugs and culture conditions known to alter their relative activity, and (233)uranium uptake was monitored. Interestingly, the inhibitory effect of colchicine, cytochalasin D, phorbol 12-myristate 13-acetate, and chlorpromazine on endocytosis was highly correlated with their effect on uranium uptake, a relationship that was not true when the NaPi-IIa transport system was studied. Whereas the competitive inhibition of the NaPi-IIa by phosphonoformic acid (PFA) significantly decreased uranium uptake, this effect was not reproduced when NaPi-IIa inhibition was mediated by the replacement of extracellular Na(+) with N-methyl-D-glucamine. Uranium uptake was also not significantly altered when NaPi-IIa expression was stimulated in MDCK cells. More surprisingly, we observed by transmission electron microscopy that uranium cytotoxicity was dependent upon the extent of its intracellular precipitation, but not on its intracellular content, and was suppressed by PFA. In conclusion, our results suggest that low-dose uranium uptake is mainly mediated by absorptive endocytosis, and we propose PFA as a potential uranium chelator.


Asunto(s)
Endocitosis , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIa/fisiología , Nitrato de Uranilo , Animales , Transporte Biológico , Supervivencia Celular/efectos de los fármacos , Relación Dosis-Respuesta a Droga , Endocitosis/efectos de los fármacos , Foscarnet/farmacología , Cinética , Células LLC-PK1 , Microscopía Electrónica de Transmisión , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIa/antagonistas & inhibidores , Proteínas Cotransportadoras de Sodio-Fosfato de Tipo IIa/metabolismo , Porcinos , Nitrato de Uranilo/metabolismo , Nitrato de Uranilo/toxicidad
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...